AAC Accepted Manuscript Posted Online 27 July 2015 Antimicrob. Agents Chemother. doi:10.1128/AAC.00899-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

The Glucose Transporter PfHT is an Antimalarial Target of the HIV Protease Inhibitor Lopinavir

2 3

Running title: HIV Drug Lopinavir Blocks Malarial Glucose Transport

4 5

Thomas E. Krafta, Christopher Armstronga, Monique R. Heitmeiera, Audrey R. Odoma,c and Paul

6

W. Hruza,b#

7 8

a

Department of Pediatrics, and bDepartment of Cell Biology and Physiology, cDepartment of

9

Molecular Microbiology, Washington University School of Medicine, St Louis MO 63110

10 11

# Address all correspondence to Paul W. Hruz.: [email protected]

12 13

1

14

Abstract

15 16

Malaria and HIV infection are co-endemic in a large portion of the world and remain a major

17

cause of morbidity and mortality. Growing resistance of Plasmodium species to existing

18

therapies has increased the need for new therapeutic approaches. The Plasmodium glucose

19

transporter PfHT is known to be essential for parasite growth and survival. We have previously

20

shown that HIV protease inhibitors (PIs) act as antagonists of mammalian glucose transporters.

21

While the PI lopinavir is known to have antimalarial activity, the mechanism-of-action is

22

unknown. We report here that lopinavir blocks glucose uptake into isolated malaria parasites

23

at therapeutically relevant drug levels. Malaria parasites depend on a constant supply of

24

glucose as their primary source of energy and decreasing the available concentration of glucose

25

leads to parasite death. We identified the malarial glucose transporter PfHT as a target for

26

inhibition by lopinavir that leads to parasite death. This discovery provides a mechanistic basis

27

for the antimalarial effect of lopinavir and provides a direct target for novel drug design with

28

utility beyond the HIV-infected population.

29 30

Introduction Despite aggressive world-wide efforts to eradicate malaria, this life-threatening disease

31 32

continues to affect over 200 million people per year, resulting in an annual death toll exceeding

33

half a million, mostly among African children(1). Currently, vaccination against malaria is not

34

available, while resistance against all known therapeutics is spreading(1). As a result, newer

35

antimalarial agents with novel mechanisms-of-action are urgently needed.

2

The global prevalence of malaria and HIV infection largely overlap geographically.

36 37

Combination antiviral therapy that includes the HIV protease inhibitor (PI) lopinavir has been

38

found to dramatically decrease malaria incidence in a pediatric clinical population by 41%,

39

suggesting a direct effect of PIs on parasite replication(2). Indeed, lopinavir has demonstrated

40

in vitro activity(3) against Plasmodium falciparum, the protozoan species responsible for most

41

malaria deaths(1, 4). In addition, lopinavir reduces the malaria liver stage burden in infected

42

rhesus monkeys in vivo at clinically relevant concentrations(5). Despite ongoing efforts, the direct cellular target(s) of lopinavir responsible for its

43 44

antimalarial properties against P. falciparum remains unclear. PIs were originally designed as

45

antagonists of the viral aspartyl protease(6). The malaria parasite requires a class of aspartyl

46

proteases called plasmepsins, which are necessary to degrade host hemoglobin(7) and direct

47

export of malaria export proteins(8); however the antimalarial activity of PIs does not appear to

48

be mediated through plasmepsin inhibition(9, 10). Identifying the antimalarial mechanism-of-

49

action of PIs is imperative for finding a novel, clinically proven drug target and developing a new

50

class of lopinavir-like antimalarial drugs. In clinical populations, prolonged use of PIs is associated with insulin resistance. Recent

51 52

studies have identified the molecular mechanism of this effect, which is mediated by direct

53

binding of PIs to the insulin-responsive facilitative glucose transporter GLUT4(11–13). The

54

human glucose transporters share sequence homology with the essential P. falciparum glucose

55

transporter, PfHT. Similar to GLUT1 and GLUT4, the predicted topology of PfHT comprises 12

56

transmembrane helices, forming a central glucose permeation path. Key residues that are

3

57

involved in glucose binding and transport are preserved between the human and malaria

58

glucose transporters(14, 15). Intraerythrocytic malaria parasites depend on a constant supply of glucose as their

59 60

primary source of energy(16). Not surprisingly, infected erythrocytes show a ~100 fold increase

61

in glucose consumption compared to uninfected erythrocytes(17). PfHT (PF3D7_0204700) is the

62

principal glucose transporter, transcribed from a single copy gene with no close paralogue(14).

63

PfHT has been genetically validated as essential in Plasmodium parasites(18) and has been

64

independently chemically validated as a novel drug target against malaria(14, 19). Here we show that lopinavir inhibits glucose uptake into the P. falciparum parasite by

65 66

blocking PfHT at therapeutically relevant concentrations. This establishes a direct molecular

67

target for the antimalarial activity of lopinavir and validates the utility of targeting PfHT in novel

68

drug development.

69 70

Material and Methods

71

Materials

72

[14C]-2-deoxy- glucose (2-DOG) was purchased from PerkinElmer. [3H]-2DOG was purchased

73

from American Radiolabels Inc. PfHT DNA was codon optimized and synthesized by Life

74

Technologies (Grand Island, NY). GLUT1 shRNA was obtained through the RNAi core at

75

Washington University, School of Medicine. HEK293 cells were acquired from the American

76

Type Culture Collection. HIV protease inhibitors were obtained through the NIH AIDS Reagent

77

Program, Division of AIDS, NIAID, NIH. Compound 3361 was kindly donated by Sanjeev Krishna

4

78

(Centre for Infection, Division of Cellular and Molecular Medicine, St. George's, University of

79

London).

80 81

Malaria tissue culture

82

The P. falciparum strain strain 3D7 was obtained from the Malaria Research and Reference

83

Reagent Resource Center (MR4, ATCC, Manassas, Virginia). Unless otherwise stated, P.

84

falciparum strains were cultured at 37°C in a 2% suspension of human erythrocytes in RPMI-

85

1640 medium (Sigma-Aldrich, SKU R4130) supplemented with 27 mM sodium bicarbonate, 11

86

mM glucose, 5 mM HEPES, 1 mM sodium pyruvate, 0.37 mM hypoxanthine, 0.01 mM

87

thymidine, 10 µg/mL gentamicin, and 0.5% Albumax (Life Technologies) in a 5% O2/ 5% CO2 /

88

90% N2 atmosphere, as previously described(20, 21). Culture growth was monitored by

89

microscopic analysis of Giemsa-stained blood smears.

90 91

Drug and glucose sensitivity of P. falciparum

92

Asynchronous P. falciparum cultures were diluted to 1% parasitemia and were treated at a

93

range of concentrations of inhibitor or glucose. Growth inhibition assays were performed in

94

opaque 96-well plates at 100 µL culture volume. After 3 days, parasite growth was quantified

95

by measuring DNA content using Picogreen (Life Technologies), as previously described(22).

96

Picogreen fluorescence was measured on a FLUOstar Omega microplate reader (BMG Labtech)

97

at 485 nm excitation and 528 nm emission. IC50 values were calculated by nonlinear regression

98

analysis using GraphPad Prism software.

99

5

100

Stable expression of PfHT

101

HEK293 cells were stably transfected with codon optimized PfHT DNA in the pcDNA 3.1(-) hygro

102

plasmid (Life Technologies) as described earlier(23). After selection with hygromycin, 10

103

colonies were grown to near confluency in 4cm tissue culture dishes and the highest expresser

104

of PfHT was selected using [3H]-2DOG uptake and quantitative RT-PCR (as described below).

105

106

Short hairpin RNA lentiviruses for GLUT1 knockdown

107

293T packaging cells were cotransfected with VSVg and Delta 8.9 packaging plasmids plus

108

shRNA lentiviral plasmids targeting GLUT1 (NM_006516.1-2310s1c1) using Optifect transfection

109

reagent (Life Technologies). The media was changed 12 hours after transfection and

110

supernatants (10 ml) were harvested every 24 hours for 72 hours and kept at 4°C until they

111

were pooled, filtered through 0.45 μm syringe filters, aliquoted and stored at -80°C until use.

112

HEK293 wild-type cells or HEK293 cells stably expressing PfHT were infected by exposing them

113

to viral supernatant for 48 hours, then replacing with selection media (containing 5 μg/ml

114

puromycin).

115

116

Quantitative PCR analysis

117

Total RNA was isolated using the TRIzol Plus RNA Purification System (Life Technologies). RNA

118

was reverse transcribed using the qScript cDNA SuperMix (Quanta Biosciences, Gaithersburg,

6

119

MD). Quantitative RT-PCR was performed using the Power SYBR® Green PCR Master Mix

120

(Applied Biosystems) as described previously(24).

121 122

2-DOG uptake into P. falciparum and HEK293 cells

123

Uptake of [14C]-2DOG into isolated parasites was determined at room temperature using the

124

methods described previously(25). Lopinavir and compound 3361 were added 5min prior to the

125

addition of [14C]-2DOG (0.2 µCi/mL final concentration). Uptake of [3H]-2DOG into HEK293 cells

126

was measured in phosphate buffer at 37°C for 5min as described previously (26).

127

128

Statistical analyses

129

The data are reported as means ±SEM. Differences between control and experimental values

130

were determined by one-way ANOVA analysis.

131 132

Modeling of PfHT and docking of lopinavir

133

PfHT structure was predicted using the I-TASSER webserver(27–29). The webserver was

134

amongst top scorers of the Server Section of the 7th (2006), 8th (2008), 9th (2010), 10th CASPs

135

(2012), and 11th CASPs (2014), a community-wide experiment for testing the state-of-the-art of

136

protein structure predictions. We chose a model that resembled the inward open

137

conformation. Docking was performed using AutoDock Vina(30). The receptor was prepared

138

using MGLTools version 1.5.6 and the ligand lopinavir was prepared using ArgusLab. The

7

139

docking process was repeated with number of modes of 50 and increasing exhaustiveness of

140

25, 50 and 100. The results in each attempt were 20 similar docked poses. Docked poses were

141

displayed using PyMOL version 1.7.4.0.

142

8

143

Results

144

We compared the effect of currently FDA-approved HIV protease inhibitors on the

145

replication and survival of P. falciparum parasites in asexual erythrocyte cell culture. Consistent

146

with prior reports(3, 9) , all tested compounds inhibited parasite growth. Lopinavir was the

147

most potent inhibitor with an IC50 of 1.9 μM (Fig. 1a). We previously determined the inhibitory

148

effect of several HIV protease inhibitors on human glucose transport(11, 13, 26). Since HIV PIs

149

inhibit human glucose transport, we evaluated whether lopinavir might inhibit glucose uptake

150

in isolated P. falciparum parasites. We compared the uptake of radiolabeled 2-DOG in live

151

parasites, isolated from erythrocytes treated with vehicle, lopinavir, or the chemically unrelated

152

PfHT inhibitor 3361 (Fig. 2a)(19). The O-3-hexose derivative compound 3361 [3-O-((undec-10-

153

en)-yl)-D-glucose)] has previously been shown to selectively inhibit PfHT relative to several

154

mammalian sugar transporters and inhibit glucose uptake by the parasite, leading to inhibition

155

of growth and proliferation in vitro and in vivo(19, 25). We found that high concentrations of

156

lopinavir potently inhibited uptake of radiolabeled glucose by more than 70% compared to

157

vehicle-treated control, similar to the effect observed by the known PfHT inhibitor compound

158

3361. In dose-response studies (Fig. 2b), we found that the half-maximal inhibitory

159

concentration (IC50) of lopinavir was 16 ± 4.2 μM, which is within the range of average serum

160

peak levels of both adult and pediatric patients treated with lopinavir/ritonavir(31–34). To confirm that inhibition of malaria glucose transport was mediated through PfHT

161 162

inhibition, we developed and tested the effect of this PI on glucose transport in a PfHT-

163

overexpressing HEK293 cell line. HEK293 cells predominantly transport glucose using the

164

facilitative transport protein GLUT1. Using a PfHT sequence codon-optimized for mammalian

9

165

cell expression, we overexpressed the malarial hexose transporter protein and knocked down

166

the expression of GLUT1 using shRNA. In this engineered cell line, PfHT was the predominant

167

hexose transporter expressed (Fig. 3a). Glucose uptake in the PfHT overexpressing cell line was

168

>5 times higher compared to background cells that did not overexpress PfHT but were

169

transduced with GLUT1 shRNA (Fig. 3b). We quantified the amount of [3H]-2-DOG accumulated

170

in PfHT overexpressing HEK293 cells in the presence of varying concentration of lopinavir and

171

determined an IC50 of 14 ± 2 μM. The IC50 values for the effect of lopinavir on glucose uptake in

172

both P. falciparum parasites and PfHT-overexpressing HEK293 cells are closely correlated,

173

consistent with a model in which inhibition of PfHT is responsible for decreased glucose uptake

174

and reduced parasite growth (Fig. 4). Supporting this model, blood stage P. falciparum is

175

acutely sensitive to reduced extracellular glucose concentrations (17). Since the measurement

176

of 2-DOG uptake involves both the transport of this sugar and phosphorylation to 2-DOG-6-P,

177

we cannot fully exclude the possibility that lopinavir also inhibits the malarial hexokinase.

178

However, the observation that lopinavir inhibits 2-DOG uptake with different IC50s when

179

measured in HEK293 cells overexpressing different GLUTs (ranging from 3μM for GLUT4 to

180

32μM for GLUT1) suggests that hexokinase is not directly targeted. To further investigate the mode of action of lopinavir on PfHT, we determined several

181 182

parameters by Michaelis-Menten kinetics. As expected, uptake of glucose is saturated with

183

increasing substrate concentration. The observed increase in Km with constant Vmax indicates

184

that lopinavir acts as a competitive inhibition of glucose uptake (Fig. 5). To investigate the binding site of lopinavir on PfHT, we generated a model of the

185 186

transporter based on several crystal structures of homologous proteins using the I-TASSER

10

187

webserver (Table 1). We selected a model with high cluster density in an inward conformation

188

to determine the lopinavir binding site by docking analysis. We created a docking volume

189

covering the entire protein to obtain unbiased results. Lopinavir was docked to the PfHT model

190

using AutoDock VINA via rigid receptor, flexible ligand docking. All docked poses of lopinavir

191

were bound to single binding pocket on the intracellular side of the glucose permeation path,

192

preventing glucose from entering and reaching its binding site (Fig. 6a). These results could be

193

replicated in several docking experiments using random seeds and different degrees of

194

exhaustiveness, strongly supporting this position as the lowest energy binding site. We

195

compared the PfHT-lopinavir putative binding site with the binding site of the structurally

196

related protease inhibitor indinavir in the PfHT homologue GLUT4, published by Hresko et al.

197

(Fig. 6b)(26). The docking experiments suggest that both indinavir and lopinavir bind to a

198

structurally similar binding pocket in their respective glucose transporters. Lopinavir binding

199

within the glucose permeation pathway is consistent with this drug acting as a competitive

200

inhibitor of zero-trans glucose uptake (Fig. 5). Although inhibition of GLUT4 by Indinavir

201

requires drug binding from the intracellular side of the transporter (35), lopinavir appears to

202

have access to this domain from the exofacial side of PfHT. Similar to the observed differences

203

in isoform-selectivity of HIV protease inhibitors for mammalian GLUTs (26), it is possible that

204

steric influences of amino acid side chains lining the glucose permeation pathway contribute to

205

the ability of these drugs to target PfHT.

206 207

11

208

Discussion

209

There is a compelling need for novel therapies for treatment of malaria. In particular,

210

the emergence and spread of resistance to artemisinin-based compounds threatens malaria

211

control efforts worldwide. Protease inhibitors, such as lopinavir, represent promising

212

therapeutic leads for new antimalarial development. Lopinavir has potent antimalarial activity

213

in vitro and in animal models(4, 5), and human studies of lopinavir-treated individuals in

214

endemic areas indicate antimalarial potency at clinically relevant doses(2). Importantly, many

215

of the pharmacologic challenges that slow antimalarial development have already been

216

overcome with lopinavir, which is orally bioavailable, available in suspension form (for pediatric

217

use), and suitable for once-daily dosing in combination with ritonavir. Our studies provide substantial evidence that inhibition of the malaria hexose

218 219

transporter, PfHT, is responsible for the antiparasitic effects of lopinavir. PfHT hexose

220

transporter function is known to be required for Plasmodium parasite development in culture

221

and in animal models(14, 18, 19, 25, 36). We find that not only does lopinavir directly block

222

glucose uptake into P. falciparum parasites, it also inhibits glucose uptake in human cells

223

engineered such that majority of hexose transport is through heterologously expressed PfHT.

224

Importantly, profound reductions of glucose uptake into parasites and into PfHT-expressing

225

human cells are achieved at lopinavir concentrations well below therapeutically achieved drug

226

levels(31–34).

227

Of note, even lower concentrations of both lopinavir and compound 3361 are required

228

to inhibit parasite replication than are required to inhibit glucose transport(19). This difference

229

may be explained by the reliance of parasites on a constant glucose supply. Plasmodium

12

230

infection increases cellular glucose consumption in erythrocytes nearly 100 fold, while parasite

231

survival is highly sensitive to reduced extracellular glucose concentrations(17). Thus, even

232

partial inhibition of Plasmodium glucose transporters may lead to growth inhibition and

233

eventually death of the parasite. PfHT may therefore represent a promising, parasite-specific

234

“Achilles Heel” for target-based drug discovery efforts. We cannot rule out the possibility that

235

there are additional targets of lopinavir in P. falciparum other than PfHT . The existence of

236

additional drug targets, if present, could delay the development of drug resistance, a major

237

problem for malaria drugs. However, observing this difference for both lopinavir and

238

compound 3361, two structurally unrelated drugs that inhibit PfHT-mediated glucose transport

239

decreases the likelihood that they would both also inhibit another secondary target. As agents designed for long-term suppression of HIV replication, lopinavir and other

240 241

protease inhibitors are well tolerated, with minor adverse events (mostly gastrointestinal)

242

associated with short-term use(37). During chronic HIV treatment regimens that include PIs,

243

disturbances in glucose homeostasis and other metabolic changes that increase cardiovascular

244

risk have been reported(38). In this HIV-infected population, it has been challenging to

245

dissociate the direct contributions of PIs from the influences of viral infection, other drug

246

exposures (e.g. nucleoside reverse transcriptase inhibitors), and associated environmental

247

factors. Nevertheless, the in vitro effects of PIs on the insulin-responsive facilitative glucose

248

transporter GLUT4 correlate directly with in vivo changes in insulin sensitivity, both in rodent

249

models(39) and in humans(40). Fortunately, the effect of PIs on glucose tolerance is readily

250

reversible following short-term drug exposure, as required for treatment of malaria(39, 40).

251

However, the full spectrum of PI-mediated effects on the remaining GLUT isoforms remains

13

252

unknown, and not all PIs appear to have the same effect on GLUT4 activity(41). Ideally, the

253

development of drugs that selectively target PfHT over mammalian GLUTs would provide highly

254

selective inhibition of PfHT over the mammalian GLUTs, resulting in a superior safety profile. The development of high-throughput screening protocols targeting the Plasmodium

255 256

glucose transporter will help identify additional classes of PfHT antagonists. Several assays have

257

been developed by expressing PfHT in yeast or Leishmania(14, 18, 42). In combination with

258

these prior studies, our current findings confirm that PfHT is an attractive, druggable target for

259

malaria. Our development of a PfHT overexpressing HEK293 cell line and demonstration that

260

this system can reliably test for drug-mediated inhibition of the malarial glucose transporter

261

can significantly aid candidate drug screening. Complementing a high-throughput approach is

262

the opportunity for structure-based design. This can allow for higher affinity and selectivity to

263

the plasmodium transporter while minimizing effects on mammalian glucose transporters

264

(GLUTs). GLUT1 is the most extensively characterized of the human GLUTs and a crystal

265

structure of this protein was recently reported(43). In the absence of structural data for PfHT

266

and the other known GLUTs, it has been necessary to rely upon molecular modeling based on

267

the structures of related bacterial and mammalian transporter. Although, our docking results

268

are consistent with the previously identified HIV protease inhibitor binding site in the closely

269

related transporter GLUT4(26), we cannot exclude the possibility that lopinavir binds to a

270

distinct binding site only accessible in an outward open conformation of PfHT. Testing this

271

possibility will generate valuable data once a homologues transporter is crystallized in the

272

outward open conformation. Future efforts to establish a comprehensive structure-activity

273

profile for the inhibition of PfHT and the human GLUTs may provide valuable information about

14

274

the feasibility of this approach. Investigation of potential additive or synergistic effects of PfHT

275

inhibitors with existing anti-malarial agents or other compounds that inhibit parasite

276

metabolism will also be of great interest. In summary, the identification of lopinavir as a clinically meaningful inhibitor of the

277 278

essential malaria hexose transporter PfHT provides a strong rationale and molecular basis for

279

future antimalarial development efforts targeting this protein. Because PfHT inhibition retards

280

both the liver and mosquito-stage development of rodent malaria parasites(44), novel PfHT

281

inhibitors hold great promise for use in malaria treatment and as transmission-blocking agents.

282

The existing pharmacokinetic and safety data for lopinavir should allow rapid assessment of the

283

suitability and efficacy of this agent in non-HIV infected patient populations. Understanding the

284

molecular target of lopinavir in P. falciparum will be key to any necessary medicinal chemical

285

optimization of lopinavir and related protease inhibitors, repurposed for use as antimalarials.

286 287

Acknowledgements

288

This work was supported by the Children’s Discovery Institute of Washington University and St.

289

Louis Children’s Hospital (MD-LI-2011-171, to A.R.O.), NIH/NIAID R01AI103280 (to A.R.O.), a

290

March of Dimes Basil O’Connor Starter Scholar Research Award (to A.R.O.), and a Doris Duke

291

Charitable Foundation Clinical Scientist Development award (to A.R.O.).

292 293

The authors thank Maria Payne for conducting quantitative PCR experiments and generating

294

plasmids.

295

15

296

References

297

1.

World Health Organization. 2014. World malaria report 2014 165–176.

298 299 300

2.

Achan J, Kakuru a, Ikilezi G, Ruel T, Clark TD, Nsanzabana C, Charlebois E, Aweeka F, Dorsey G, Rosenthal PJ, Havlir D, Kamya MR. 2012. Antiretroviral agents and prevention of malaria in HIV-infected Ugandan children. N Engl J Med 367:2110–2118.

301 302

3.

Nsanzabana C, Rosenthal PJ. 2011. In vitro activity of antiretroviral drugs against Plasmodium falciparum. Antimicrob Agents Chemother 55:5073–5077.

303 304 305

4.

Hobbs C V, Voza T, Coppi A, Kirmse B, Marsh K, Borkowsky W, Sinnis P. 2009. HIV protease inhibitors inhibit the development of preerythrocytic-stage plasmodium parasites. J Infect Dis 199:134–41.

306 307 308 309

5.

Hobbs C V., Neal J, Conteh S, Donnelly L, Chen J, Marsh K, Lambert L, Orr-Gonzalez S, Hinderer J, Healy S, Borkowsky W, Penzak SR, Chakravarty S, Hoffman SL, Duffy PE. 2014. HIV treatments reduce malaria liver stage burden in a non-human primate model of malaria infection at clinically relevant concentrations in vivo. PLoS One 9:e100138.

310 311 312

6.

Nalam MNL, Schiffer CA. 2008. New approaches to HIV protease inhibitor drug design II: testing the substrate envelope hypothesis to avoid drug resistance and discover robust inhibitors. Curr Opin HIV AIDS 3:642–6.

313 314 315

7.

Banerjee R, Liu J, Beatty W, Pelosof L, Klemba M, Goldberg DE. 2002. Four plasmepsins are active in the Plasmodium falciparum food vacuole, including a protease with an active-site histidine. Proc Natl Acad Sci 99:990–995.

316 317 318

8.

Boddey JA, Hodder AN, Günther S, Gilson PR, Patsiouras H, Kapp EA, Pearce JA, de Koning-Ward TF, Simpson RJ, Crabb BS, Cowman AF. 2010. An aspartyl protease directs malaria effector proteins to the host cell. Nature 463:627–631.

319 320 321

9.

Parikh S, Liu J, Sijwali P, Gut J, Goldberg DE, Rosenthal PJ. 2006. Antimalarial effects of human immunodeficiency virus type 1 protease inhibitors differ from those of the aspartic protease inhibitor pepstatin. Antimicrob Agents Chemother 50:2207–2209.

322 323 324

10.

Hobbs C V, Tanaka TQ, Muratova O, Van Vliet J, Borkowsky W, Williamson KC, Duffy PE. 2013. HIV treatments have malaria gametocyte killing and transmission blocking activity. J Infect Dis 208:139–48.

325 326 327

11.

Hresko RC, Hruz PW. 2011. HIV Protease Inhibitors Act as Competitive Inhibitors of the Cytoplasmic Glucose Binding Site of GLUTs with Differing Affinities for GLUT1 and GLUT4. PLoS One 6:e25237.

16

328 329

12.

Murata H, Hruz PW, Mueckler M. 2000. The mechanism of insulin resistance caused by HIV protease inhibitor therapy. J Biol Chem 275:20251–20254.

330 331

13.

Murata H, Hruz PW, Mueckler M. 2002. Indinavir inhibits the glucose transporter isoform Glut4 at physiologic concentrations. AIDS 16:859–863.

332 333

14.

Slavic K, Krishna S, Derbyshire ET, Staines HM. 2011. Plasmodial sugar transporters as anti-malarial drug targets and comparisons with other protozoa. Malar J 10:165.

334 335

15.

Woodrow CJ, Penny JI, Krishna S. 1999. Intraerythrocytic Plasmodium falciparum expresses a high affinity facilitative hexose transporter. J Biol Chem 274:7272–7.

336 337

16.

Kirk K. 2001. Membrane transport in the malaria-infected erythrocyte. Physiol Rev 81:495–537.

338 339

17.

Mehta M, Sonawat HM, Sharma S. 2006. Glycolysis in Plasmodium falciparum results in modulation of host enzyme activities 95–103.

340 341 342 343

18.

Blume M, Hliscs M, Rodriguez-Contreras D, Sanchez M, Landfear S, Lucius R, Matuschewski K, Gupta N. 2011. A constitutive pan-hexose permease for the Plasmodium life cycle and transgenic models for screening of antimalarial sugar analogs. FASEB J 25:1218–29.

344 345

19.

Joet T, Eckstein-Ludwig U, Morin C, Krishna S. 2003. Validation of the hexose transporter of Plasmodium falciparum as a novel drug target. Proc Natl Acad Sci U S A 100:7476–9.

346 347 348

20.

Zhang B, Watts KM, Hodge D, Kemp LM, Hunstad DA, Hicks LM, Odom AR. 2011. A second target of the antimalarial and antibacterial agent fosmidomycin revealed by cellular metabolic profiling. Biochemistry 50:3570–7.

349 350

21.

Trager W, Jensen JB. 1976. Human malaria parasites in continuous culture. Science 193:673–5.

351 352 353

22.

Corbett Y, Herrera L, Gonzalez J, Cubilla L, Capson TL, Coley PD, Kursar TA, Romero LI, Ortega-Barria E. 2004. A novel DNA-based microfluorimetric method to evaluate antimalarial drug activity. Am J Trop Med Hyg 70:119–24.

354 355 356

23.

Chaudhary S, Pak JE, Gruswitz F, Sharma V, Stroud RM. 2012. Overexpressing human membrane proteins in stably transfected and clonal human embryonic kidney 293S cells. Nat Protoc 7:453–66.

357 358 359

24.

Aerni-Flessner L, Abi-Jaoude M, Koenig A, Payne M, Hruz PW. 2012. GLUT4, GLUT1, and GLUT8 are the dominant GLUT transcripts expressed in the murine left ventricle. Cardiovasc Diabetol 11:63.

17

360 361 362

25.

Saliba KJ, Krishna S, Kirk K. 2004. Inhibition of hexose transport and abrogation of pH homeostasis in the intraerythrocytic malaria parasite by an O-3-hexose derivative. FEBS Lett 570:93–96.

363 364 365

26.

Hresko RC, Kraft TE, Tzekov A, Wildman S a., Hruz PW. 2014. Isoform-selective inhibition of facilitative glucose transporters: Elucidation of the molecular mechanism of HIV protease inhibitor binding. J Biol Chem 289:16100–16113.

366 367

27.

Zhang Y. 2008. I-TASSER server for protein 3D structure prediction. BMC Bioinformatics 9:40.

368 369

28.

Roy A, Kucukural A, Zhang Y. 2010. I-TASSER: a unified platform for automated protein structure and function prediction. Nat Protoc 5:725–738.

370 371

29.

Yang J, Yan R, Roy A, Xu D, Poisson J, Zhang Y. 2014. The I-TASSER Suite: protein structure and function prediction. Nat Methods 12:7–8.

372 373 374

30.

Trott O, Olson AJ. 2010. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 31:455–61.

375 376 377 378

31.

Bergshoeff AS, Fraaij PL, Ndagijimana J, Verweel G, Hartwig NG, Niehues T, De Groot R, Burger DM. 2005. Increased dose of lopinavir/ritonavir compensates for efavirenzinduced drug-drug interaction in HIV-1-infected children. J Acquir Immune Defic Syndr 39:63–68.

379 380 381 382

32.

Rosso R, Di Biagio A, Dentone C, Gattinara GC, Martino AM, Viganò A, Merlo M, Giaquinto C, Rampon O, Bassetti M, Gatti G, Viscoli C. 2006. Lopinavir/ritonavir exposure in treatment-naive HIV-infected children following twice or once daily administration. J Antimicrob Chemother 57:1168–1171.

383 384 385

33.

Best BM, Stek AM, Mirochnick M, Hu C, Li H, Burchett SK, Rossi SS, Smith E, Read JS, Capparelli E V. 2010. Lopinavir tablet pharmacokinetics with an increased dose during pregnancy. J Acquir Immune Defic Syndr 54:381–388.

386 387 388 389

34.

Murphy RL, Brun S, Hicks C, Eron JJ, Gulick R, King M, White a C, Benson C, Thompson M, Kessler H a, Hammer S, Bertz R, Hsu a, Japour a, Sun E. 2001. ABT-378/ritonavir plus stavudine and lamivudine for the treatment of antiretroviral-naive adults with HIV-1 infection: 48-week results. AIDS 15:F1–F9.

390 391 392

35.

Hresko RC, Hruz PW. 2011. HIV Protease Inhibitors Act as Competitive Inhibitors of the Cytoplasmic Glucose Binding Site of GLUTs with Differing Affinities for GLUT1 and GLUT4. PLoS One 6:e25237.

18

393 394 395

36.

Slavic K, Straschil U, Reininger L, Doerig C, Morin C, Tewari R, Krishna S. 2010. Life cycle studies of the hexose transporter of Plasmodium species and genetic validation of their essentiality. Mol Microbiol 75:1402–13.

396 397

37.

Chandwani A, Shuter J. 2008. Lopinavir / ritonavir in the treatment of HIV-1 infection : a review 4:1023–1033.

398 399 400

38.

Grunfeld C, Kotler DP, Arnett DK, Falutz JM, Haffner SM, Hruz P, Masur H, Meigs JB, Mulligan K, Reiss P, Samaras K. 2008. Contribution of metabolic and anthropometric abnormalities to cardiovascular disease risk factors. Circulation 118:e20–8.

401 402

39.

Hruz PW, Murata H, Qiu H, Mueckler M. 2002. Indinavir Induces Acute and Reversible Peripheral Insulin Resistance in Rats. Diabetes 51:937–942.

403 404

40.

Noor MA, Lo JC, Mulligan K, Schwarz JM, Halvorsen RA, Schambelan M, Grunfeld C. 2001. Metabolic effects of indinavir in healthy HIV-seronegative men. AIDS 15:F11–8.

405 406 407

41.

Noor MA, Flint OP, Maa J-F, Parker RA. 2006. Effects of atazanavir/ritonavir and lopinavir/ritonavir on glucose uptake and insulin sensitivity: demonstrable differences in vitro and clinically. AIDS 20:1813–1821.

408 409 410 411

42.

Ortiz D, Guiguemde WA, Johnson A, Elya C, Anderson J, Clark J, Connelly M, Yang L, Min J, Sato Y, Guy RK, Landfear SM. 2015. Identification of Selective Inhibitors of the Plasmodium falciparum Hexose Transporter PfHT by Screening Focused Libraries of AntiMalarial Compounds. PLoS One 10:e0123598.

412 413

43.

Deng D, Xu C, Sun P, Wu J, Yan C, Hu M, Yan N. 2014. Crystal structure of the human glucose transporter GLUT1. Nature 510:121–5.

414 415 416 417

44.

Slavic K, Delves MJ, Prudêncio M, Talman AM, Straschil U, Derbyshire ET, Xu Z, Sinden RE, Mota MM, Morin C, Tewari R, Krishna S, Staines HM. 2011. Use of a selective inhibitor to define the chemotherapeutic potential of the plasmodial hexose transporter in different stages of the parasite’s life cycle. Antimicrob Agents Chemother 55:2824–30.

418 419

19

420

Author Contributions

421

T.E.K. and P.W.H. conceived the initial idea of the study. T.E.K., C.A. and M.H. performed the

422

experiments. T.E.K. created the model and performed the docking. T.E.K., A.R.O. and P.W.H.

423

contributed to the experimental design and data analysis. T.E.K., A.R.O. and P.W.H. wrote the

424

manuscript with feedback from M.H.

425 426

Competing Financial Interests statement

427

The authors declare no competing financial interests.

428 429

20

430

Figure Legends

431 432

Figure 1: Comparison of HIV protease inhibitors’ potency on P. falciparum growth inhibition (a)

433

IC50 comparison of HIV protease inhibitors on growth P. falciparum in blood culture. IC50 was

434

calculated using non-linear regression analysis and is expressed as means with 95% confidence

435

interval. (b) Chemical structures of the three most potent inhibitors HIV protease inhibitors of

436

PfHT mediated glucose uptake.

437 438

Figure 2: (a) Uptake of [14C]2DOG by isolated P. falciparum trophozoites in the presence of

439

lopinavir (100 μM), compound 3361 (200 μM) or vehicle. The parasites were suspended in

440

HEPES-buffered saline containing 200 μM glucose. (b) Uptake of [14C]2DOG by isolated P.

441

falciparum trophozoites at increasing concentrations of lopinavir. The uptake was quenched

442

after 2 minutes. IC50 was calculated using non-linear regression analysis. Uptake data are

443

expressed as means ±SEM. A 30sec or 1min time point would have been a more accurate

444

measurement of initial rates however, we chose the longer time point to enhance the uptake

445

signal as the degree of non-linearity will probably not greatly affect the calculated IC50 value.

446 447

Figure 3. A. mRNA expression levels of glucose transporter proteins in HEK293 wild-type (WT),

448

GLUT1 knock-down (KD) and GLUT1 KD + PfHT overexpressing (OE) cells. mRNA levels were

449

determined via quantitative PCR and levels are displayed in copy number per ng of cDNA. Data

450

are expressed as means ±SEM. B. Comparing specific activity of glucose uptake in HEK293 cells

21

451

expressing GLUT1 shRNA and plus and minus overexpression of PfHT. Data are expressed as

452

means ±SEM (*P

The Glucose Transporter PfHT1 Is an Antimalarial Target of the HIV Protease Inhibitor Lopinavir.

Malaria and HIV infection are coendemic in a large portion of the world and remain a major cause of morbidity and mortality. Growing resistance of Pla...
1KB Sizes 1 Downloads 7 Views